Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Pain Res Manag ; 2023: 8893932, 2023.
Article in English | MEDLINE | ID: mdl-38047157

ABSTRACT

Background: Premature infants often undergo painful procedures and consequently experience repeated procedural neonatal pain. This can elicit hyperalgesia and cognitive impairment in adulthood. Treatments for neonatal pain are limited. Orientin is a flavonoid C-glycoside that has repeatedly been shown to have pharmacological effects in the past decades. The aim of this study was to systematically explore the effect of orientin on repeated procedural neonatal pain using network pharmacology, molecular docking analysis, and experimental validation. Methods: Several compound-protein databases and disease-protein databases were employed to identify proteins that were both predicted targets of orientin and involved in neonatal pain. A protein-protein interaction (PPI) network was constructed, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed to explore the potential mechanism of action. Molecular docking analysis was employed to calculate the binding energy and visualize the interactions between orientin and potential target proteins. Finally, a mouse model of repeated procedural neonatal pain was established and orientin was administered for 6 days. The mechanical and thermal pain thresholds were assessed in neonates and adult mice. A Morris water maze was employed to investigate cognitive impairment in adult mice. Results: A total of 286 proteins that were both predicted targets of orientin and involved in neonatal pain were identified. The hub proteins were SRC, HSP90AA1, MAPK1, RHOA, EGFR, AKT1, PTPN11, ESR1, RXRA, and HRAS. GO analysis indicated that the primary biological process (BP), molecular function (MF), and cellular component (CC) were protein phosphorylation, protein kinase activity, and vesicle lumen, respectively. KEGG analysis revealed that the mitogen-activated protein kinase (MAPK) signaling pathway may be the key to the mechanism of action. Molecular docking analysis showed the high binding affinities of orientin for MAPK1, MAPK8, and MAPK14. In mice, orientin inhibited the hyperalgesia in the pain threshold tests in neonates and adult mice and cognitive impairment in adult mice. Immunofluorescence showed that phosphorylated MAPK1 (p-ERK) protein levels in the hippocampus and spinal dorsal horn were downregulated by orientin. Conclusion: The findings suggested that orientin alleviates neonatal pain, and the MAPK signaling pathway is involved.


Subject(s)
Hyperalgesia , Pain, Procedural , Humans , Adult , Infant , Animals , Mice , Molecular Docking Simulation , Hyperalgesia/drug therapy , Network Pharmacology , Flavonoids , Pain
2.
BMB Rep ; 53(12): 658-663, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33298240

ABSTRACT

The N-myc downstream regulated gene (NDRG) family members are dysregulated in several tumors. Functionally, NDRGs play an important role in the malignant progression of cancer cells. However, little is known about the potential implications of NDRG4 in pancreatic ductal adenocarcinoma (PDAC). The aim of the current study was to elucidate the expression pattern of NDRG4 in PDAC and evaluate its potential cellular biological effects. Here, we firstly report that epigenetic-mediated silencing of NDRG4 promotes PDAC by regulating mitochondrial function. Data mining demonstrated that NDRG4 was significantly down-regulated in PDAC tissues and cells. PDAC patients with low NDRG4 expression showed poor prognosis. Epigenetic regulation by DNA methylation was closely associated with NDRG4 down-regulation. NDRG4 overexpression dramatically suppressed PDAC cell growth and metastasis. Further functional analysis demonstrated that up-regulated NDRG4 in SW1990 and Canpan1 cells resulted in attenuated mitochondrial function, including reduced ATP production, decreased mitochondrial membrane potential, and increased fragmented mitochondria. However, opposite results were obtained for HPNE cells with NDRG4 knockdown. These results indicate that hypermethylation-driven silencing of NDRG4 can promote PDAC by regulating mitochondrial function and that NDRG4 could be as a potential biomarker for PDAC patients. [BMB Reports 2020; 53(12): 658-663].


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Muscle Proteins/metabolism , Nerve Tissue Proteins/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/physiopathology , Apoptosis/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/physiopathology , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , China , DNA Methylation/genetics , Databases, Genetic , Epigenesis, Genetic/genetics , Epithelial-Mesenchymal Transition/physiology , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Mitochondria/metabolism , Muscle Proteins/genetics , Neoplasm Invasiveness/genetics , Nerve Tissue Proteins/genetics
3.
J Oncol ; 2020: 8847687, 2020.
Article in English | MEDLINE | ID: mdl-33204264

ABSTRACT

Reprogrammed glucose metabolism of enhanced aerobic glycolysis, also known as Warburg effect, which exerts a significant contributor to cancer progression, is regarded as a hallmark of cancer. The roles of long noncoding RNAs (lncRNA) in regulating cancer via metabolic reprogramming are mostly unknown, including esophagal cancer (EC). Here, we showed that how the lncRNA urothelial carcinoma associated 1 (UCA1) exerts pro-oncogene in regulating EC glucose metabolism. Firstly, we found that upregulated UCA1 expression enhances the malignant phenotypes of EC, including poor outcome, larger tumor size, positive lymphatic invasion, and advanced pathological stages. UCA1 silencing could suppress EC cell proliferation and metastasis. Following, bioinformatics analyses revealed that UCA1 regulated the HK2 expression through functioning as a competing endogenous RNA (ceRNA). Mechanistically, UCA1 overexpression could elevate the activation of HK2 oncogenes via inhibition of miR-203 activity, as evidenced by the positive correlation of UCA1 with HK2 and inverse correlation with miR-203 expression. Luciferase activity assay further verified the targeting relationship between UCA1, miR-203, and HK2. Upregulated UCA1 in EC cells significantly suppressed the degradation of HK2 by miR-203. Further research showed that upregulated UCA1 effectively increased the rate of glucose uptake, lactate output, and ECAR value, all of which can be attenuate by HK2 interference and 2-DG, whereas knockdown of UCA1 had the opposite effect. In sum, our findings suggest that the UCA1/miR-203/HK2 axis contributes to EC development by reprogramming tumor glucose metabolism, providing new insight into the management of EC patients.

4.
Cell Death Dis ; 11(4): 291, 2020 04 27.
Article in English | MEDLINE | ID: mdl-32341354

ABSTRACT

Leukemic stem cells (LSCs) and hematopoietic stem cells (HSCs) are both dependent on the hypoxic bone marrow (BM) microenvironment (also known as the BM niche). There is always fierce competition between the two types of cells, and the former exhibits a greater competitive advantage than the latter via multiple mechanisms. Under hypoxia, the dynamic balance between the generation and clearing of intracellular reactive oxygen species (ROS) is conducive to maintaining a quiescent state of cells. Quiescent LSCs can reside well in the BM niche, avoiding attack by chemotherapeutic agents, which is the cause of chemotherapeutic resistance and relapse in leukemia. HSCs acquire energy mainly through anaerobic glycolysis, whereas LSCs achieve energy metabolism largely through mitochondrial oxidative respiration. Mitochondria are the primary site of ROS generation. Thus, in theory, mitochondria-mediated respiration will cause an increase in ROS generation in LSCs and a higher intracellular oxidative stress level. The sensitivity of the cells to pro-oxidant drugs increases as well, which allows for the selective clearing of LSCs by pro-oxidative therapy. However, HSCs are also highly sensitive to changes in ROS levels, and the toxic effects of pro-oxidant drugs on HSCs poses a major challenge to pro-oxidative therapy in leukemia. Given the above facts, we reviewed studies on the oxidative resistance of LSCs and the oxidative damage to HSCs under pro-oxidative therapy. An in-depth investigation into the oxidative stress status and regulatory mechanisms of LSCs and HSCs in hypoxic environments will promote our understanding of the survival strategy employed by LSCs and the mechanism of the oxidative damage to HSCs in the BM niche, thus facilitating individualized treatment of leukemia patients and helping eliminate LSCs without disturbing normal hematopoietic cells.


Subject(s)
Hematopoietic Stem Cells/metabolism , Leukemia/genetics , Oxidative Stress/genetics , Cell Differentiation , Humans , Leukemia/pathology , Tumor Microenvironment
5.
Gene ; 741: 144552, 2020 May 30.
Article in English | MEDLINE | ID: mdl-32165297

ABSTRACT

Hypoxia, as a form of stress, plays a critical role in oncogenesis, including metabolic reprogramming. Mitochondrial, the centers of energy production, re-balance mitochondria dynamic to maintain cell survival during high levels of environmental stresses. NDRG1 is a hypoxia-inducible protein that is involved in various human cancers, including HCC. However, little is known about whether NDRG1 participants in the quality control of mitochondrial in times of stress. Here, we firstly showed that how NDRG1 exerted its role through mediating mitochondrial dynamic in HCC cells under hypoxia. Initially, we identified that NDRG1 expression varies with oxygen content. NDRG1 silencing notably induced cell apoptosis under hypoxia, while no obviously change of wildtype cells in hypoxia compared with that in normoxia. Further analysis revealed that NDRG1 silencing in HCC cells led to increase of pro apoptotic protein BAX and decrease in anti-apoptotic proteins Bcl-2 and Bclx, which meant mitochondrial damage were induced. In the analysis of mitochondria, we found that more released cytochrome c located in cytosolic with NDRG1 knockdown in hypoxia, which may be due to mitochondria division. And the following experiment proved that more fragmented mitochondria were presented in NDRG1 silencing cells, as well as destroyed mitochondrial membrane potential with evidence by JC-1 was verified. Moreover, these trends could be reversed by Mdivi1. Further research showed that NDRG1 silencing disrupt hypoxia-enhanced aerobic glycolysis through effectively decreased glucose uptake, lactate output and ECAR value. In sum, we provide the first direct evidence that NDRG1-driven change in mitochondrial dynamics and aerobic glycolysis maintain cells survival in HCC during hypoxia.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Cycle Proteins/genetics , Cellular Reprogramming/genetics , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/genetics , Apoptosis/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Cytochromes c/genetics , Gene Expression Regulation, Neoplastic/genetics , Glycolysis/genetics , Humans , Liver Neoplasms/pathology , Membrane Potential, Mitochondrial/genetics , Mitochondrial Dynamics/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Hypoxia/genetics , bcl-X Protein/genetics
6.
Curr Drug Targets ; 21(5): 477-498, 2020.
Article in English | MEDLINE | ID: mdl-31736443

ABSTRACT

Reactive oxygen species (ROS), an important molecule inducing oxidative stress in organisms, play a key role in tumorigenesis, tumor progression and recurrence. Recent findings on ROS have shown that ROS can be used to treat cancer as they accelerate the death of tumor cells. At present, pro-oxidant drugs that are intended to increase ROS levels of the tumor cells have been widely used in the clinic. However, ROS are a double-edged sword in the treatment of tumors. High levels of ROS induce not only the death of tumor cells but also oxidative damage to normal cells, especially bone marrow hemopoietic cells, which leads to bone marrow suppression and (or) other side effects, weak efficacy of tumor treatment and even threatening patients' life. How to enhance the killing effect of ROS on tumor cells while avoiding oxidative damage to the normal cells has become an urgent issue. This study is a review of the latest progress in the role of ROS-mediated programmed death in tumor treatment and prevention and treatment of oxidative damage in bone marrow induced by ROS.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Marrow/drug effects , Hematopoiesis/drug effects , Neoplasms/drug therapy , Reactive Oxygen Species/pharmacology , Antineoplastic Agents/therapeutic use , Bone Marrow Cells , Oxidative Stress/drug effects , Reactive Oxygen Species/therapeutic use
7.
Biochem Biophys Res Commun ; 511(2): 323-329, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30782485

ABSTRACT

Mitochondrial Ca2+ uptake, an important governing for Ca2+ homeostasis, is catalyzed by the mitochondrial calcium uniporter (MCU) complex. SMDT1, as a subunit of MCU complex, was essential for bridging the calcium-sensing role of MICU1 and MICU2 with the calcium-conducting role of MCU. However, the molecular mechanism and regulatory purpose of SMDT1 remain largely unexplored, especially no study was reported in cancer. Here, we firstly reported that how SMDT1 exerted its role through mediating mitochondrial dynamic in PDAC malignancy. In this study, by screening online of subunit of MCU complex, we confirmed that SMDT1 expression was significantly positive correlated with PDAC prognosis. The GEO datasets showed decreased SMDT1 expression in PDAC tumor compared with non-tumor tissues. SMDT1 overexpression could notably inhibit cell proliferation and induce cell apoptosis. Further analysis demonstrated that up-regulated SMDT1 in ASPC1 and Canpan1 cells led to increased accumulation of pro apoptotic protein BAX and decrease in anti-apoptotic proteins Bcl-2 and Bclx. And more releasing of cytochrome c located in cytosolic. Mechanistically, in the morphological analysis of mitochondria, more fragmented mitochondria were presented in SMDT1 overexpression cells by promting the phosphorylation of Drp1, increasing Fis and decreasing MFN1. Meanwhile, more Drp1 was translocated on the mitochondrial from the cytoplasm in up-regulated SMDT1 cells. On the basis of the evidence above we deduce that SMDT1-driven change in mitochondrial dynamics mediated cells apoptosis in PDAC. And, SMDT1 could serve as an important therapeutic target to normalize mitochondrial dynamic responsible for poor prognosis in PDAC.


Subject(s)
Calcium Channels/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Mitochondrial Dynamics , Pancreatic Neoplasms/metabolism , Apoptosis , Calcium Channels/analysis , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation , Humans , Mitochondria/metabolism , Mitochondria/pathology , Pancreatic Neoplasms/pathology
8.
Int Immunopharmacol ; 58: 72-79, 2018 May.
Article in English | MEDLINE | ID: mdl-29558662

ABSTRACT

Neuropathic pain affects patients worldwide. The therapeutic effects of current methods are still poor. This study was performed to investigate the neuro-protective effect of orientin in rats with spinal nerve ligation (SNL). In this study, the paw mechanical withdrawal threshold (PWT) and the paw thermal withdrawal latency (PWL) behavioral assays indicated that orientin alleviated the warm and mechanical allodynia in rats with SNL. The enzyme-linked immunosorbent assay (ELISA) showed that orientin suppressed the levels of pro-inflammatory cytokines interleukin-6 (IL-6), interleukin-1ß (IL-1ß), and tumor necrosis factor alpha (TNF-α) and increased the levels of anti-inflammatory cytokine interleukin-10 (IL-10). Malondialdehyde (MDA) levels were down-regulated while superoxide dismutase (SOD) and glutathione (GSH) levels were up-regulated by orientin. OX42 and GFAP immune fluorescent staining results demonstrated that orientin inhibited the activation of microglia and astrocytes in rats with SNL. Western blot analysis indicated that the neuroprotective effect of orientin was mediated by inhibition of Toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-kappa B) signaling pathway. This study suggested that orientin is a promising neuroprotective agent suitable for therapy for neuropathic pain.


Subject(s)
Astrocytes/physiology , Flavonoids/therapeutic use , Glucosides/therapeutic use , Microglia/physiology , Neuralgia/drug therapy , Neuroprotective Agents/therapeutic use , Postoperative Complications/drug therapy , Animals , Behavior, Animal , Cytokines/metabolism , Disease Models, Animal , Humans , Inflammation Mediators/metabolism , Ligation , NF-kappa B/metabolism , Neuralgia/etiology , Poaceae/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Spinal Nerves/surgery , Toll-Like Receptor 4/metabolism
9.
Int J Clin Exp Pathol ; 11(2): 568-576, 2018.
Article in English | MEDLINE | ID: mdl-31938142

ABSTRACT

Receptor-interacting serine-threonine kinase 3 (RIPk3) is a key signaling molecule in the regulation of cell apoptosis and necroptosis, it plays an important role in the pathophysiological changes of many hematologic diseases. However, the regulatory role of RIPk3 in programmed cell death (PCD) is not fully known. In this study, bone marrow-specific RIPk3 gene knockout homozygotes (RIPk3-/- mice) were established by homologous recombination. The physiological index of peripheral blood, the morphology and structure of the bone marrow, the bone marrow nucleated cells (BMNCs), the hemopoietic stem cells (HSCs), interleukin-6 (IL-6) level and the colony formation capacity of bone marrow hematopoietic progenitor cells were compared between RIPk3-/- mice and wild-type mice. The results showed that, the cell death rate of BMNCs in RIPk3-/- mice was significantly higher than that in control mice, indicated that RIPk3 gene knockout may cause damage to bone marrow cells to some extent. However, the bone marrow had normal structure and morphology in the bone marrow-specific RIPk3-knockout mice, and there were not significantly different between the two mice in most of the blood physiological indicators, and colony yields of hemopoietic stem/progenitor cells. Further study found that the bone marrow IL-6 level of the RIPk3-/- mice increased significantly, besides, the number of BMNCs and HSCs in the bone marrow of the RIPk3-/- mice increased considerably as compared with the control mice. The findings implies that bone marrow RIPk3 gene knockout may lead to the increase of BMNCs cell death, however, increased secretion of hematopoietic cytokines such as IL-6 may promote the proliferation of hematopoietic stem/progenitor cells and thus maintain the stability of bone marrow hematopoiesis. This hypothesis and the detailed mechanisms remain to be further investigated.

10.
Neurochem Res ; 43(2): 375-386, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29119332

ABSTRACT

Vinorine is a monoterpenoid indole alkaloid, a type of natural alkaloids. Growing reports exhibited the numerous pharmacology activities of vinorine such as anti-inflammation, anti-bacterial and anti-tumor. In this study, the effect of vinorine injection (7.5, 15 and 30 mg/kg) on motor function, sensation and nerve regeneration in sciatic nerve crush injury rat was investigated. The results of behavioral analysis, electrophysiological analysis and muscle histological analysis suggested that vinorine promoted the motor function recovery after sciatic nerve injury. The results of mechanical withdrawal thresholds assay and hot plate test demonstrated that vinorine improved the sensation recovery after sciatic nerve injury. The results of Fluoro-gold retrograde labeling, transmission electron microscope assay, toluidine blue and HE staining showed that vinorine attenuated the nerve damage caused by sciatic nerve injury and promoted the nerve regeneration. Furthermore, nerve growth factor (NGF) and its downstream extracellular signal-regulated kinase (ERK) signaling pathway participated in the neuro-recovery effect of vinorine after crush. In conclusion, vinorine treatment accelerated the sciatic nerve regeneration, motor function recovery and sensation recovery after crush injury via regulation of NGF and ERK activity. These results suggested that vinorine is a promising agent for never injury therapy.


Subject(s)
Indole Alkaloids/pharmacology , Recovery of Function/drug effects , Sciatic Nerve/drug effects , Sciatic Neuropathy/drug therapy , Animals , Disease Models, Animal , Indole Alkaloids/chemistry , Male , Nerve Crush/methods , Nerve Regeneration/drug effects , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/pathology , Rats, Sprague-Dawley , Sciatic Nerve/injuries
11.
Med Sci Monit ; 22: 2301-8, 2016 Jul 02.
Article in English | MEDLINE | ID: mdl-27371503

ABSTRACT

BACKGROUND 7, 8, 3'-trihydroxyflavone (THF) is a novel pro-neuronal small molecule that acts as a TrkB agonist. In this study, we examined the effect of THF on promoting neuronal growth and protecting anesthetics-induced neurotoxicity in dorsal root ganglion (DRG) neurons in vitro. MATERIAL AND METHODS Neonatal mouse DRG neurons were cultured in vitro and treated with various concentrations of THF. The effect of THF on neuronal growth was investigated by neurite outgrowth assay and Western blot. In addition, the protective effects of THF on bupivacaine-induced neurotoxicity were investigated by apoptosis TUNEL assay, neurite outgrowth assay, and Western blot, respectively. RESULTS THF promoted neurite outgrowth of DRG neurons in dose-dependent manner, with an EC50 concentration of 67.4 nM. Western blot analysis showed THF activated TrkB signaling pathway by inducing TrkB phosphorylation. THF also rescued bupivacaine-induced neurotoxicity by reducing apoptosis and protecting neurite retraction in DRG neurons. Furthermore, the protection of THF in bupivacaine-injured neurotoxicity was directly associated with TrkB phosphorylation in a concentration-dependent manner in DRG neurons. CONCLUSIONS THF has pro-neuronal effect on DRG neurons by promoting neurite growth and protecting against bupivacaine-induced neurotoxicity, likely through TrkB activation.


Subject(s)
Bupivacaine/toxicity , Flavones/pharmacology , Ganglia, Spinal/drug effects , Neurites/drug effects , Neuronal Outgrowth/drug effects , Neurons/drug effects , Animals , Apoptosis/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Mice , Mice, Inbred C57BL , Neurites/metabolism , Neurites/pathology , Neurons/metabolism , Neurons/pathology , Protective Agents/pharmacology , Receptor, trkB/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...